FGFR/PD-1 combination therapy for the treatment of cancer
Provided herein are combination therapies for the treatment of cancer. In particular, the disclosed methods are directed to treatment of cancer in a patient comprising administering an antibody that blocks the interaction between PD-1 and PD-L1 and an FGFR inhibitor, wherein the antibody that blocks the interaction between PD-1 and PD-L1 and the FGFR inhibitor are administered if one or more FGFR variants are present in a biological sample from the patient.
Latest ASTEX THERAPEUTICS LTD Patents:
- Combination of an FGFR inhibitor and a CMET inhibitor
- PHARMACEUTICAL COMPOSITIONS COMPRISING N-(3,5-DIMETHOXYPHENYL)-N'-(1-METHYLETHYL)-N-[3-(1-METHYL-1H-PYRAZOL-4-YL)QUINOXALIN-6-YL]ETHANE-1,2-DIAMINE
- Pharmaceutical compositions comprising N-(3,5-dimethoxyphenyl)-N?-(1-methylethyl)-N-[3-(1-methyl-1H-pyrazol-4-yl)quinoxalin-6-yl]ethane-1,2-diamine
- FGFR/PD-1 COMBINATION THERAPY FOR THE TREATMENT OF CANCER
- PYRAZOLYL QUINOXALINE KINASE INHIBITORS
This application claims priority to U.S. Provisional Application No. 62/142,569, filed Apr. 3, 2015, the disclosure of which is hereby incorporated by reference in its entirety.
SEQUENCE LISTINGThe instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Mar. 22, 2016, is named PRD3366USNP_SL.txt and is 53,086 bytes in size.
TECHNICAL FIELDProvided herein are combination therapies for the treatment of cancer. In particular, the disclosed methods are directed to treatment of cancer in a patient comprising administering an antibody that blocks the interaction between PD-1 and PD-L1 and a fibroblast growth factor receptor (FGFR) inhibitor.
BACKGROUNDFor cancer patients failing the main therapeutic option (front-line therapy) for that cancer type, there is often no accepted standard of care for second and subsequent-line therapy, unless a particular genetic abnormality is identified and a specific therapy is available. Fibroblast growth factor receptors (FGFRs) are a family of receptor tyrosine kinases involved in regulating cell survival, proliferation, migration and differentiation. FGFR alterations have been observed in some cancers. To date, there are no approved therapies that are efficacious in patients with FGFR alterations.
SUMMARYDisclosed herein are methods of using a combination therapy comprising an antibody that blocks the interaction between PD-1 and PD-L1 and an FGFR inhibitor to treat cancer in the patient. In some embodiments, the methods comprise administering to the patient a pharmaceutically effective amount of an antibody that blocks the interaction between PD-1 and PD-L1 and a pharmaceutically effective amount of an FGFR inhibitor, wherein the antibody that blocks the interaction between PD-1 and PD-L1 and the FGFR inhibitor are administered if one or more FGFR variants are present in a biological sample from the patient.
In other embodiments, the methods of treating cancer in a patient comprise: administering to the patient a pharmaceutically effective amount of an antibody that blocks the interaction between PD-1 and PD-L1; monitoring the efficacy of the antibody; and, if the antibody is not efficacious, evaluating a biological sample from the patient for a presence of one or more FGFR variants and administering to the patient a pharmaceutically effective amount of an FGFR inhibitor if the one or more FGFR variants are present in the sample.
The summary, as well as the following detailed description, is further understood when read in conjunction with the appended drawings. For the purpose of illustrating the disclosed methods, there are shown in the drawings exemplary embodiments of the methods; however, the methods are not limited to the specific embodiments disclosed. In the drawings:
The disclosed methods may be understood more readily by reference to the following detailed description taken in connection with the accompanying figures, which form a part of this disclosure. It is to be understood that the disclosed methods are not limited to the specific methods described and/or shown herein, and that the terminology used herein is for the purpose of describing particular embodiments by way of example only and is not intended to be limiting of the claimed methods.
Unless specifically stated otherwise, any description as to a possible mechanism or mode of action or reason for improvement is meant to be illustrative only, and the disclosed methods are not to be constrained by the correctness or incorrectness of any such suggested mechanism or mode of action or reason for improvement.
Reference to a particular numerical value includes at least that particular value, unless the context clearly dictates otherwise. When a range of values is expressed, another embodiment includes from the one particular value and/or to the other particular value. Further, reference to values stated in ranges include each and every value within that range. All ranges are inclusive and combinable.
When values are expressed as approximations, by use of the antecedent “about,” it will be understood that the particular value forms another embodiment.
The term “about” when used in reference to numerical ranges, cutoffs, or specific values is used to indicate that the recited values may vary by up to as much as 10% from the listed value. Thus, the term “about” is used to encompass variations of ±10% or less, variations of ±5% or less, variations of ±1% or less, variations of ±0.5% or less, or variations of ±0.1% or less from the specified value.
It is to be appreciated that certain features of the disclosed methods which are, for clarity, described herein in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the disclosed methods that are, for brevity, described in the context of a single embodiment, may also be provided separately or in any subcombination.
As used herein, the singular forms “a,” “an,” and “the” include the plural.
The following abbreviations are used throughout the disclosure: FFPE (formalin-fixed, paraffin-embedded); NSCLC (non-small-cell lung carcinoma); SCLC (small-cell lung cancer); FGFR (fibroblast growth factor receptor); PD-1 (programmed cell death 1); PD-L1 (programmed death-ligand 1); FGFR3:TACC3 (fusion between genes encoding FGFR3 and transforming acidic coiled-coil containing protein); FGFR3:BAIAP2L1 (fusion between genes encoding FGFR3 and brain-specific angiogenesis inhibitor 1-associated protein 2-like protein 1); FGFR2:AFF3 (fusion between genes encoding FGFR2 and AF4/FMR2 family, member 3); FGFR2:BICC1 (fusion between genes encoding FGFR2 and bicaudal C homolog 1); FGFR2:CASP7 (fusion between genes encoding FGFR2 and caspase 7); FGFR2:CCDC6 (fusion between genes encoding FGFR2 and coiled-coil domain containing 6); FGFR2:OFD1 (fusion between genes encoding FGFR2 and oral-facial-digital syndrome 1).
The term “antibody” refers to (a) immunoglobulin polypeptides, i.e., polypeptides of the immunoglobulin family that contain an antigen binding site that specifically binds to a specific antigen (e.g., PD-1 or PD-L1), including all immunoglobulin isotypes (IgG, IgA, IgE, IgM, IgD, and IgY), classes (e.g. IgG1, IgG2, IgG3, IgG4, IgA1, IgA2), subclasses, and various monomeric and polymeric forms of each isotype, unless otherwise specified, and (b) conservatively substituted variants of such immunoglobulin polypeptides that immunospecifically bind to the antigen (e.g., PD-1 or PD-L1). Antibodies are generally described in, for example, Harlow & Lane, Antibodies: A Laboratory Manual (Cold Spring Harbor Laboratory Press, 1988). Unless otherwise apparent from the context, reference to an antibody also includes antibody derivatives as described in more detail below.
“Antibody fragments” comprise a portion of a full length antibody, generally the antigen-binding or variable region thereof, such as Fab, Fab′, F(ab′)2, and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; and multispecific antibodies formed from antibody fragments. Various techniques have been developed for the production of antibody fragments, including proteolytic digestion of antibodies and recombinant production in host cells; however, other techniques for the production of antibody fragments will be apparent to the skilled practitioner. In some embodiments, the antibody fragment of choice is a single chain Fv fragment (scFv). “Single-chain Fv” or “scFv” antibody fragments comprise the VH and VL domains of antibody, wherein these domains are present in a single polypeptide chain. Generally, the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the scFv to form the desired structure for antigen binding. For a review of scFv and other antibody fragments, see James D. Marks, Antibody Engineering, Chapter 2, Oxford University Press (1995) (Carl K. Borrebaeck, Ed.).
An “antibody derivative” means an antibody, as defined above, that is modified by covalent attachment of a heterologous molecule such as, e.g., by attachment of a heterologous polypeptide (e.g., a cytotoxin) or therapeutic agent (e.g., a chemotherapeutic agent), or by glycosylation, deglycosylation, acetylation or phosphorylation not normally associated with the antibody, and the like.
The term “monoclonal antibody” refers to an antibody that is derived from a single cell clone, including any eukaryotic or prokaryotic cell clone, or a phage clone, and not the method by which it is produced. Thus, the term “monoclonal antibody” is not limited to antibodies produced through hybridoma technology.
“Biological sample” refers to any sample from a patient in which cancerous cells can be obtained and protein expression can be evaluated and/or RNA can be isolated. Suitable biological samples include, but are not limited to, blood, lymph fluid, bone marrow, sputum, a solid tumor sample, or any combination thereof. In some embodiments, the biological sample can be formalin-fixed paraffin-embedded tissue (FFPET).
As used here, “block(s) the interaction” refers to the ability of an anti-PD-1 antibody or an anti-PD-L1 antibody to inhibit or reduce binding of PD-L1 to PD-1, such that signaling/functioning through PD-1 is abolished or diminished.
As used herein, “FGFR variant” refers to an alteration in the wild type FGFR gene, including, but not limited to, FGFR fusion genes, FGFR mutations, FGFR amplifications, or any combination thereof “FGFR fusion” or “FGFR fusion gene” refers to a gene encoding a portion of FGFR (e.g., FGRF2 or FGFR3) and one of the herein disclosed fusion partners created by a translocation between the two genes.
As used herein, “patient” is intended to mean any animal, in particular, mammals. Thus, the methods are applicable to human and nonhuman animals, although most preferably with humans. “Patient” and “subject” may be used interchangeably herein.
“Pharmaceutically effective amount” refers to an amount of an antibody that blocks the interaction between PD-1 and PD-L1 and an amount of an FGFR inhibitor that treats the patient.
As used herein, “treating” and like terms refer to reducing the severity and/or frequency of cancer symptoms, eliminating cancer symptoms and/or the underlying cause of said symptoms, reducing the frequency or likelihood of cancer symptoms and/or their underlying cause, and improving or remediating damage caused, directly or indirectly, by cancer.
Disclosed herein are methods of treating cancer in a patient comprising: administering to the patient a pharmaceutically effective amount of an antibody that blocks the interaction between PD-1 and PD-L1 and a pharmaceutically effective amount of an FGFR inhibitor, wherein the antibody that blocks the interaction between PD-1 and PD-L1 and the FGFR inhibitor are administered if one or more FGFR variants are present in a biological sample from the patient.
PD-1 is a cell surface receptor expressed on the surface of CD4+ and CD8+ T cells, B cells, and myeloid cells. The ligands of PD-1, PD-L1 and PD-L2, are expressed on immune cells; in addition, PD-L1 is also expressed on cancer cells. When engaged by its ligands, PD-1 downregulates the immune response by reducing T cell proliferation, cytokine production and effector function. Antibodies against PD-1 (anti-PD-1 antibodies) and/or its ligands (anti-PD-L1 antibodies, for example) can block the interaction between PD-1 and PD-L1, thereby inhibiting the downregulation of the immune response. The disclosed methods comprise administering to the patient a pharmaceutically effective amount of an antibody that blocks the interaction between PD-1 and PD-L1. In some embodiments, the methods can comprise administering to the patient a pharmaceutically effective amount of an anti-PD-1 antibody. In some embodiments, the methods can comprise administering to the patient a pharmaceutically effective amount of an anti-PD-L1 antibody. In some embodiments, the methods can comprise administering to the patient a pharmaceutically effective amount of an anti-PD-1 antibody and an anti-PD-L1 antibody.
Exemplary anti-PD-1 antibodies include, but are not limited to, OPDIVO® (nivolumab) (Bristol-Myers Squibb) and KEYTRUDA® (pembrolizumab) (Merck). Exemplary anti-PD-L1 antibodies include, but are not limited to, MPDL3208A (Roche) and MEDI4736 (AstraZeneca).
Exemplary FGFR inhibitors are described in U.S. Publ. No. 2013/0072457 A1 (incorporated herein by reference) and include N-(3,5-dimethoxyphenyl)-N′-(1-methylethyl)-N-[3-(1-methyl-1H-pyrazol-4-yl)quinoxalin-6-yl]ethane-1,2-diamine (referred to herein “JNJ-42756493”), including any tautomeric or stereochemically isomeric forms thereof, N-oxides thereof, pharmaceutically acceptable salts thereof, or solvates thereof (suitable R groups are also disclosed in U.S. Publ. No. 2013/0072457 A1). Thus, in some embodiments, the FGFR inhibitor can be the compound of formula (I):
or a pharmaceutically acceptable salt thereof. In some aspects, the pharmaceutically acceptable salt is a HCl salt.
The antibody that blocks the interaction between PD-1 and PD-L1 and the FGFR inhibitor can be administered as a single therapeutic agent or can be co-administered as individual agents. When administered as individual agents, the antibody and FGFR inhibitor can be administered contemporaneously or sequentially in either order. In some embodiments, the antibody that blocks the interaction between PD-1 and PD-L1 and the FGFR inhibitor can be administered contemporaneously. In some embodiments, the antibody that blocks the interaction between PD-1 and PD-L1 can be administered sequentially. In some aspects, for example, the antibody that blocks the interaction between PD-1 and PD-L1 can be administered first, followed by administration of the FGFR inhibitor. In other aspects, the FGFR inhibitor can be administered first, followed by administration of the antibody that blocks the interaction between PD-1 and PD-L1. When administered sequentially, the antibody and FGFR inhibitor can be administered within seconds, minutes, hours, days, or weeks of each other.
The pharmaceutically effective amount of the antibody that blocks the interaction between PD-1 and PD-L1 and FGFR inhibitor will be dependent on several factors including, but not limited to, stage and severity of the cancer, as well as other factors relating to the health of the patient. Those skilled in the art would know how to determine the pharmaceutically effective amount.
The disclosed methods are suitable for treating cancer in a patient if one or more FGFR variants are present in a biological sample from the patient. In some embodiments, the FGFR variant can be one or more FGFR fusion genes. In some embodiments, the FGFR variant can be one or more FGFR mutations. In some embodiments, the FGFR variant can be one or more FGFR amplifications. In some embodiments, a combination of the one or more FGFR variants can be present in the biological sample from the patient. For example, in some embodiments, the FGFR variants can be one or more FGFR fusion genes and one or more FGFR mutations. In some embodiments, the FGFR variants can be one or more FGFR fusion genes and one or more FGFR amplifications. In some embodiments, the FGFR variants can be one or more FGFR mutations and one or more FGFR amplifications. In yet other embodiments, the FGFR variants can be one or more FGFR fusion genes, mutations, and amplifications.
Exemplary FGFR fusion genes are provided in Table 1 and include, but are not limited to: FGFR2:AFF3; FGFR2:BICC1; FGFR2:CASP7; FGFR2:CCDC6; FGFR2:OFD1; FGFR3:BAIAP2L1; FGFR3:TACC3-Intron; FGFR3:TACC3V1; FGFR3:TACC3V3; or a combination thereof. The sequences of the FGFR fusion genes are disclosed in Table 6.
The methods can further comprise evaluating the presence of one or more FGFR variants in the biological sample before the administering step. Suitable methods for evaluating a biological sample for the presence of one or more FGFR variants are disclosed elsewhere herein.
The disclosed methods can be dependent upon PD-L1 expression in the cancer or can be carried out irrespectively of PD-L1 expression in the cancer. In some embodiments, for example, the methods can comprise administering to the patient a pharmaceutically effective amount of an antibody that blocks the interaction between PD-1 and PD-L1 and a pharmaceutically effective amount of an FGFR inhibitor, wherein the antibody that blocks the interaction between PD-1 and PD-L1 and the FGFR inhibitor are administered if one or more FGFR variants are present in a biological sample from the patient and PD-L1 expression in the biological sample from the patient is at a specified level or within a specified range. In some aspects, for example, the methods can be carried out if the PD-L1 expression is high in the biological sample. Accordingly, in some embodiments the methods can comprise administering to the patient a pharmaceutically effective amount of an antibody that blocks the interaction between PD-1 and PD-L1 and a pharmaceutically effective amount of an FGFR inhibitor, wherein the antibody that blocks the interaction between PD-1 and PD-L1 and the FGFR inhibitor are administered if PD-L1 expression is high and one or more FGFR variants are present in a biological sample from the patient. Alternatively, the methods can be carried out if the PD-L1 expression is low in the biological sample. Accordingly, the methods can comprise administering to the patient a pharmaceutically effective amount of an antibody that blocks the interaction between PD-1 and PD-L1 and a pharmaceutically effective amount of an FGFR inhibitor, wherein the antibody that blocks the interaction between PD-1 and PD-L1 and the FGFR inhibitor are administered if PD-L1 expression is low and one or more FGFR variants are present in a biological sample from the patient. The methods can be carried out if the PD-L1 expression is moderate. Accordingly, the methods can comprise administering to the patient a pharmaceutically effective amount of an antibody that blocks the interaction between PD-1 and PD-L1 and a pharmaceutically effective amount of an FGFR inhibitor, wherein the antibody that blocks the interaction between PD-1 and PD-L1 and the FGFR inhibitor are administered if PD-L1 expression is moderate and one or more FGFR variants are present in a biological sample from the patient. As discussed elsewhere herein, PD-L1 expression levels can be based upon a numerical H-score (low includes an H-score of about 0 to about 99; moderate includes an H-score of about 100 to about 199; and high includes an H-score of about 200 to about 300) or can be based upon a comparison to a reference value.
In other embodiments, the methods can be carried out irrespectively of PD-L1 expression in the biological sample from the patient and can be based on the presence of one or more FGFR variants without factoring in PD-L1 expression.
The methods can further comprise evaluating PD-L1 expression in the biological sample from the patient. Exemplary methods of evaluating PD-L1 expression are disclosed elsewhere herein. PD-L1 expression can be evaluated before, during, or after the administering step.
In some embodiments, the methods can comprise evaluating the presence of one or more FGFR variants and PD-L1 expression in the biological sample from the patient before the administering step.
Suitable biological samples evaluating PD-L1 expression, evaluating the presence of one or more FGFR variants, or for evaluating both PD-L1 expression and the presence of one or more FGFR variants include, but are not limited to, blood, lymph fluid, bone marrow, a solid tumor sample, or any combination thereof.
The disclosed methods can be used to treat a variety of cancer types including, but not limited to, lung cancer, bladder cancer, gastric cancer, breast cancer, ovarian cancer, head and neck cancer, esophageal cancer, glioblastoma, or any combination thereof. In some embodiments, the methods can be used to treat lung cancer. The lung cancer can be non-small cell lung cancer (NSCLC) adenocarcinoma, NSCLC squamous cell carcinoma, small cell lung cancer, or any combination thereof. Thus, in some aspects, the methods can be used to treat NSCLC adenocarcinoma. In other aspects, the methods can be used to treat NSCLC squamous cell carcinoma. In yet other aspects, the methods can be used to treat small cell lung cancer. In some embodiments, the methods can be used to treat bladder cancer. In some embodiments, the methods can be used to treat gastric cancer. In some embodiments, the methods can be used to treat breast cancer. In some embodiments, the methods can be used to treat ovarian cancer. In some embodiments, the methods can be used to treat head and neck cancer. In some embodiments, the methods can be used to treat esophageal cancer. In some embodiments, the methods can be used to treat glioblastoma. In some embodiments, the methods can be used to treat any combination of the above cancers.
Also disclosed are methods of treating cancer in a patient comprising: administering to the patient a pharmaceutically effective amount of an antibody that blocks the interaction between PD-1 and PD-L1; monitoring the efficacy of the antibody; and if the antibody is not efficacious, evaluating a biological sample from the patient for a presence of one or more FGFR variants and administering to the patient a pharmaceutically effective amount of an FGFR inhibitor if the one or more FGFR variants are present in the sample.
The efficacy of the antibody can be monitored by, for example, evaluating the patient's symptoms for progression of the cancer, evaluating the severity of the cancer symptoms, evaluating the frequency of the cancer symptoms, measuring tumor size, or any combination thereof. Without intent to be limiting, progression or failure to reduce the progression of the cancer, increased severity or no change in severity of the cancer symptoms, increased frequency or no change in the frequency of the cancer symptoms, increased size or no change in size of the tumor, or any combination thereof, can be indications that the antibody is not efficacious.
In some embodiments, the methods can comprise administering to the patient a pharmaceutically effective amount of an anti-PD-1 antibody. In some embodiments, the methods can comprise administering to the patient a pharmaceutically effective amount of an anti-PD-L1 antibody. In some embodiments, the methods can comprise administering to the patient a pharmaceutically effective amount of an anti-PD-1 antibody and an anti-PD-L1 antibody. Exemplary anti-PD-1 antibodies include, but are not limited to, OPDIVO® (nivolumab) (Bristol-Myers Squibb) and KEYTRUDA® (pembrolizumab) (Merck). Exemplary anti-PD-L1 antibodies include, but are not limited to, MPDL3208A (Roche) and MEDI4736 (AstraZeneca).
Exemplary FGFR inhibitors include those disclosed above, including N-(3,5-dimethoxyphenyl)-N′-(1-methylethyl)-N-[3-(1-methyl-1H-pyrazol-4-yl)quinoxalin-6-yl]ethane-1,2-diamine (referred to herein “JNJ-42756493”), including any tautomeric or stereochemically isomeric forms thereof, N-oxides thereof, pharmaceutically acceptable salts thereof, or solvates thereof (suitable R groups are also disclosed in U.S. Publ. No. 2013/0072457 A1). In some embodiments, the FGFR inhibitor can be the compound of formula (I):
or a pharmaceutically acceptable salt thereof. In some aspects, the pharmaceutically acceptable salt is a HCl salt.
The pharmaceutically effective amount of the antibody and FGFR inhibitor will be dependent on several factors including, but not limited to, stage and severity of the cancer, as well as other factors relating to the health of the patient. Those skilled in the art would know how to determine the pharmaceutically effective amount.
The disclosed methods are suitable for treating cancer in a patient if one or more FGFR variants are present in a biological sample from the patient. In some embodiments, the FGFR variant can be one or more FGFR fusion genes. In some embodiments, the FGFR variant can be one or more FGFR mutations. In some embodiments, the FGFR variant can be one or more FGFR amplifications. In some embodiments, a combination of the one or more FGFR variants can be present in the biological sample from the patient. For example, in some embodiments, the FGFR variants can be one or more FGFR fusion genes and one or more FGFR mutations. In some embodiments, the FGFR variants can be one or more FGFR fusion genes and one or more FGFR amplifications. In some embodiments, the FGFR variants can be one or more FGFR mutations and one or more FGFR amplifications. In yet other embodiments, the FGFR variants can be one or more FGFR fusion genes, mutations, and amplifications. Exemplary FGFR fusion genes are provided in Table 1 and include, but are not limited to: FGFR2:AFF3; FGFR2:BICC1; FGFR2:CASP7; FGFR2:CCDC6; FGFR2:OFD1; FGFR3:BAIAP2L1; FGFR3:TACC3-Intron; FGFR3:TACC3V1; FGFR3:TACC3V3; or a combination thereof.
Suitable methods for evaluating a biological sample for the presence of one or more FGFR variants are disclosed elsewhere herein.
The disclosed methods can be dependent upon PD-L1 expression in the biological sample or can be carried out irrespectively of PD-L1 expression in the cancer. In some aspects, for example, if the antibody is not efficacious, the methods can comprise measuring an expression level of PD-L1 in the biological sample and administering to the patient a pharmaceutically effective amount of an FGFR inhibitor if the PD-L1 expression is at a specified level or within a specified range. Methods of evaluating PD-L1 expression are disclosed elsewhere herein. The methods can be carried out if the PD-1 expression in the biological sample is low. In some embodiments, for example, the evaluating step can further comprise measuring an expression level of PD-L1 in the biological sample and the second administering step can comprise administering the FGFR inhibitor if the expression level of PD-L1 is low. In some aspects, methods of treating cancer in a patient comprise: administering to the patient a pharmaceutically effective amount of an antibody that blocks the interaction between PD-1 and PD-L1; monitoring the efficacy of the antibody; and if the antibody is not efficacious, evaluating a biological sample from the patient for a presence of one or more FGFR variants and measuring an expression level of PD-L1 in the biological sample, and administering to the patient a pharmaceutically effective amount of an FGFR inhibitor if the one or more FGFR variants are present and if the expression level of PD-L1 is low in the sample.
The methods can be carried out if the PD-1 expression in the biological sample is moderate. Thus, the evaluating step can further comprise measuring an expression level of PD-L1 in the biological sample and the second administering step can comprise administering the FGFR inhibitor if the expression level of PD-L1 is moderate. The methods can be carried out if the PD-1 expression in the biological sample is high. For example, the evaluating step can further comprise measuring an expression level of PD-L1 in the biological sample and the second administering step can comprise administering the FGFR inhibitor if the expression level of PD-L1 is high.
As discussed elsewhere herein, PD-L1 expression levels can be based upon a numerical H-score (low includes an H-score of about 0 to about 99; moderate includes an H-score of about 100 to about 199; and high includes an H-score of about 200 to about 300) or can be based upon a comparison to a reference value.
In other embodiments, the methods can be carried out irrespectively of PD-L1 expression in the cancer and can be based on the presence of one or more FGFR variants in the biological sample without factoring in PD-L1 expression.
Suitable biological samples include, but are not limited to, blood, lymph fluid, bone marrow, a solid tumor sample, or any combination thereof.
The disclosed methods can be used to treat a variety of cancer types including, but not limited to, lung cancer, bladder cancer, gastric cancer, breast cancer, ovarian cancer, head and neck cancer, esophageal cancer, glioblastoma, or any combination thereof. In some embodiments, the methods can be used to treat lung cancer. The lung cancer can be non-small cell lung cancer (NSCLC) adenocarcinoma, NSCLC squamous cell carcinoma, small cell lung cancer, or any combination thereof. Thus, in some aspects, the methods can be used to treat NSCLC adenocarcinoma. In other aspects, the methods can be used to treat NSCLC squamous cell carcinoma. In yet other aspects, the methods can be used to treat small cell lung cancer. In some embodiments, the methods can be used to treat bladder cancer. In some embodiments, the methods can be used to treat gastric cancer. In some embodiments, the methods can be used to treat breast cancer. In some embodiments, the methods can be used to treat ovarian cancer. In some embodiments, the methods can be used to treat head and neck cancer. In some embodiments, the methods can be used to treat esophageal cancer. In some embodiments, the methods can be used to treat glioblastoma. In some embodiments, the methods can be used to treat any combination of the above cancers.
Evaluating a Sample for the Presence of One or More FGFR Variants
The following methods for evaluating a biological sample for the presence of one or more FGFR variants apply equally to any of the above disclosed methods of treatment.
Suitable methods for evaluating a biological sample for the presence of one or more FGFR variants are described in the methods section herein and in U.S. Provisional Patent App. No. 62/056,159, which is incorporated herein in its entirety. For example, and without intent to be limiting, evaluating a biological sample for the presence of one or more FGFR variants can comprise any combination of the following steps: isolating RNA from the biological sample; synthesizing cDNA from the RNA; and amplifying the cDNA (preamplified or non-preamplified). In some embodiments, evaluating a biological sample for the presence of one or more FGFR variants can comprise: amplifying cDNA from the patient with a pair of primers that bind to and amplify one or more FGFR variants; and determining whether the one or more FGFR variants are present in the sample. In some aspects, the cDNA can be pre-amplified. In some aspects, the evaluating step can comprise isolating RNA from the sample, synthesizing cDNA from the isolated RNA, and pre-amplifying the cDNA.
Suitable primer pairs for performing an amplification step include, but are not limited to, those disclosed in U.S. Provisional Patent App. No. 62/056,159, as exemplified below:
The presence of one or more FGFR variants can be evaluated at any suitable time point including upon diagnosis, following tumor resection, following first-line therapy, during clinical treatment, or any combination thereof.
Evaluating PD-L1 Expression in the Cancer
The following methods for evaluating PD-L1 expression in a biological sample apply equally to any of the above disclosed methods of treatment.
In some embodiments, the disclosed methods can be dependent upon PD-L1 expression in the biological sample from the patient. Thus, administering to the patient a pharmaceutically effective amount of an antibody that blocks the interaction between PD-1 and PD-L1 and a pharmaceutically effective amount of an FGFR inhibitor may be based upon PD-L1 expression and the presence of one or more FGFR variants in the biological sample from the patient. The methods can comprise evaluating PD-L1 expression in a biological sample from the patient. The biological sample from which PD-L1 expression is evaluated can be the same biological sample from which the presence of one or more FGFR variants are evaluated, or the biological samples from which PD-L1 expression is evaluated can be a different biological sample from which the presence of one or more FGFR variants are evaluated. “Same biological sample” refers to a single sample from which both PD-L1 expression and FGFR variants are evaluated. “Different biological sample” includes the same source of sample (blood, lymph fluid, bone marrow, a solid tumor sample, etc.) taken at different time points or different sources of sample. For example, a blood sample can be obtained from the patient, evaluated for PD-L1 expression or the presence of one or more FGFR variants, and at a later time point, another blood sample can be obtained from the patient and evaluated for the presence of one or more FGFR variants or PD-L1 expression. Conversely, a blood sample can be obtained from the patient and evaluated for PD-L1 expression and/or the presence of one or more FGFR variants and a solid tumor sample can be obtained from the patient and evaluated for the presence of one or more FGFR variants and/or PD-L1 expression.
In some embodiments, the level of PD-L1 expression can be converted into a numerical H-score (as described in the methods section herein). The level of PD-L1 expression can be converted into a numerical H-score of: low PD-L1 expression, which includes an H-score of about 0 to about 99; moderate PD-L1 expression, which includes an H-score of about 100 to about 199; or high PD-L1 expression, which includes an H-score of about 200 to about 300. Treating the patient can be based upon these H-scores. For example, if the treatment methods are carried out on a patient with a low H-score, that patient would have PD-L1 expression corresponding to an H-score of about 0 to about 99. If the treatment methods are carried out on a patient with a moderate H-score, that patient would have PD-L1 expression corresponding to an H-score of about 100 to about 199. If the treatment methods are carried out on a patient with a high H-score, that patient would have PD-L1 expression corresponding to an H-score of about 200 to about 300.
In other embodiments, the level of PD-L1 expression can be compared to a reference PD-L1 expression level. In a preferred embodiment, the reference PD-L1 expression level can be predetermined. For example, a reference data set may be established using samples from unrelated patients with low, moderate and high PD-L1 expression levels. This data set can represent a standard by which relative PD-L1 expression levels are compared among patients and/or quantified using the H-Score method. In some embodiments, the reference PD-L1 expression level can be determined by comparing a patient population that is administered the antibody that blocks the interaction between PD-1 and PD-L1 to a patient population that is administered placebo. The PD-L1 expression level for each patient in the respective populations can be determined in accordance with the methods described herein. Clinical outcomes (e.g., progression-free survival or overall survival) for the patient populations can be monitored. Clinical outcomes for the patient populations relative to PD-L1 expression levels can then be compared. The reference PD-L1 expression level can correspond to the PD-L1 expression level above which the patient population that is administered the antibody that blocks the interaction between PD-1 and PD-L1 demonstrates a statistically significant improvement in at least one clinical outcome relative to the patient population that is administered placebo. A patient PD-L1 expression level that is less than the reference PD-L1 expression level, particularly when combined with the presence of one or more FGFR variants in a patient sample, can be indicative that the patient will benefit from treatment with the antibody that that blocks the interaction between PD-1 and PD-L1 in combination with an FGFR inhibitor. For example, in some embodiments, the methods can comprise administering an antibody that blocks the interaction between PD-1 and PD-L1 and an FGFR inhibitor, wherein the antibody that blocks the interaction between PD-1 and PD-L1 and the FGFR inhibitor are administered if one or more FGFR variants are present in a biological sample from the patient and the PD-L1 expression in the biological sample is less than a reference PD-L1 expression level, wherein the reference PD-L1 expression level corresponds to a PD-L1 expression level above which treatment with the antibody that blocks the interaction between PD-1 and PD-L1 alone is likely to be efficacious.
Methods for determining PD-L1 expression include, but are not limited to, immunohistochemistry (IHC), Western Blotting, microscopy, immunoprecipitation, BCA assays, spectrophotometry, or any combination thereof. Exemplary methods for evaluating PD-L1 expression are described in the methods section herein.
PD-L1 expression can be evaluated at any suitable time point including upon diagnosis, following tumor resection, following first-line therapy, during clinical treatment, or any combination thereof.
The following examples are provided to further describe some of the embodiments disclosed herein. The examples are intended to illustrate, not to limit, the disclosed embodiments.
EXAMPLESMethods
PD-L1 Immunohistochemistry
PD-L1 immunohistochemistry (IHC) was performed at a CRO (QualTek, Newtown, Pa.). Samples were stained using a CD274 PD-L1 (RUO) assay. Slides stained with a CD274 PD-L1 (RUO) assay were examined in random order and/or in blinded fashion by a board-certified clinical pathologist, the Medical Director of QualTek Clinical Laboratories (CAP/CLIA facility). The entire tissue section was evaluated for CD274 PD-L1. Only viable tissue was evaluated; areas of necrosis or obviously poorly fixed areas of tissue were not evaluated.
The tumor H-Score was calculated from the intensity of CD274 PD-L1 membrane reactivity on a four-point semi-quantitative scale (0: null, negative or non-specific staining of cell membranes; 1+: low or weak intensity staining of cell membranes; 2+: medium or moderate intensity staining of cell membranes; and 3+: high or strong intensity staining of cell membranes) and the estimated percentage of CD274 PD-L1 positive tumor cells (0-100%) for each discrete intensity value.
Tumor CD274 PD-L1 membrane reactivity was captured by a standard H-Score−the tumor H-Score minimum of 0 and the tumor H-Score maximum of 300: Tumor H-Score=([% positive cells at 1+]*1)+([% positive cells at 2+]*2)+([% positive cells at 3+]*3)
Next-Generation Sequencing (NGS)
NGS for FGFR mutations and gene amplification was performed by Foundation Medicine, Cambridge, Mass. using the FoundationOne panel (http://www.foundationmedicine.com).
FGFR Fusions
FGFR fusions were determined using a proprietary qRT-PCR assay developed by Janssen Oncology Translational Research as described in U.S. Provisional Application No. 62/056,159.
Results
PD-L1 Expression in Tumors with FGFR Fusions and Mutations
To determine the overlap of PD-L1 expression with FGFR alterations, immunohistochemistry (IHC) for PD-L1 was performed on human tumor tissue samples which were subsequently assessed for FGFR alterations. FGFR amplifications and mutations were identified using next-generation sequencing (Foundation Medicine panel, FMI). FGFR fusions were screened for using a Janssen-developed qRT-PCR assay.
Correlation of FGFR Mutations and Amplification with PD-L1
PD-L1 expression was first assessed in a set of 120 commercially sourced lung FFPE tumor tissues comprised of forty of each of the following lung tumor histologies; non-small-cell lung carcinoma (NSCLC) adenocarcinoma; NSCLC squamous cell carcinoma; and small-cell lung cancer (SCLC). FGFR mutations and gene amplification were detected using the Foundation Medicine panel. PD-L1 staining versus FGFR status was plotted for each tumor type (
FGFR Fusions and PD-L1 Expression in Bladder and NSCLC
The set of 120 lung FFPE tumor tissues was subsequently screened for FGFR fusions using a Janssen-developed qRT-PCR assay (as described in U.S. Provisional Application No. 62/056,159) detecting nine fusions (Table 1). Results for PD-L1 expression by FGFR fusion status for the NSCLC tumor samples (n=80) are shown in
Forty-five commercially sourced bladder tumors were sequenced for mutations by the Foundation Medicine panel (FMI), stained for PD-L1 expression, and screened for FGFR gene fusions using the Janssen qRT-PCR assay. Forty-two of 45 samples (93%) were positive for FGFR fusions. Five samples (11%) were positive for an FGFR mutation (FGFR3-R248C or FGFR3-S249C), all of which were also positive for FGFR fusions. PD-L1 staining H-scores for samples with FGFR alterations are summarized in Table 4, and listed in Table 5. For FGFR fusion positive samples, 22/37 (59%) were negative for PD-L1 staining. Ten FGFR fusion-positive samples (27%) expressed low levels of PD-L1, and five samples (14%) showed high PD-L1 expression. All samples with both FGFR mutations and FGFR fusions in the same tumor sample (n=5) were negative for PD-L1 staining. Overall, PD-L1 staining was absent in 64% (27/42) of bladder samples with FGFR alterations, keeping in mind that almost all of the tumors in this sample set were positive for FGFR fusions.
FGFR mutation and PD-L1 expression data were available for seven commercially sourced metastatic NSCLC samples with FGFR fusions (Janssen). No PD-L1 staining was observed in 4/7 (57%) of samples. Two samples exhibited very low PD-L1 staining, H-scores of 4 and 15. One sample showed moderate PD-L1 with an H-score of 160. Interestingly, the FGFR fusion-positive sample with moderate PD-L1 staining harbored an FGFR4 V550I mutation—an FGFR gatekeeper residue mutation with potential to confer resistance to tyrosine kinase inhibitors.
Overall these data show that the majority of commercially available tumor samples harboring FGFR alterations have very little expression or do not express PD-L1.
FGFR in Vitro Experiments
To determine the effects of JNJ427564493 on immune cell viability in vitro, peripheral blood mononuclear cells (PBMCs) from normal donors were stimulated with anti-CD3 antibodies to activate T cells, in the presence of increasing concentrations of JNJ42756493. Unstimulated PBMCs were also included to determine if JNJ42756493 affected unactivated immune populations. Cell viability was assessed at four different time points, over 6 days.
JNJ42756493 was next tested to analyze the impact on the activity of anti-PD-1 antibodies in two in vitro functional assays: Mixed Lymphocyte Reaction (MLR); and Cytomegalovirus antigen assay (CMV). For the MLR assay, CD4+ T cells are stimulated with allogeneic dendritic cells, leading to T cell activation and IFN-γ secretion. In this assay, anti-PD-1 antibodies caused dose-dependent increases in IFN-γ levels (
In the CMV assay, PBMCs from CMV-reactive donors were stimulated by the addition of CMV antigen. CMV-reactive T cells are active, expand and secrete pro-inflammatory cytokines such as IFN-γ. In the presence of anti-PD-1 antibodies, significantly higher levels of IFN-γ were secreted upon CMV stimulation (
Those skilled in the art will appreciate that numerous changes and modifications can be made to the preferred embodiments and that such changes and modifications can be made without departing from the spirit of the invention. It is, therefore, intended that the appended claims cover all such equivalent variations as fall within the true spirit and scope of the invention.
The disclosures of each patent, patent application, and publication cited or described in this document are hereby incorporated herein by reference, in its entirety.
Nucleotide Sequence of FGFR Fusion Genes
The nucleotide sequences for the FGFR fusion cDNA are provided in Table 6. The underlined sequences correspond to either FGFR3 or FGFR2, the sequences in black represent the fusion partners and the sequence in italic fonts represent the intron sequence of the FGFR3 gene.
Claims
1. A method of treating cancer in a patient comprising:
- administering to the patient a pharmaceutically effective amount of an antibody that blocks the interaction between PD-1 and PD-L1;
- monitoring the efficacy of the antibody; and
- if the antibody is not efficacious, evaluating a biological sample from the patient for a presence of one or more FGFR variants, wherein the one or more FGFR variants comprise an FGFR fusion gene, and wherein the FGFR fusion gene is FGFR2:AFF3; FGFR2:BICC1; FGFR2:CASP7; FGFR2:CCDC6; FGFR2:OFD1; FGFR3:BAIAP2L1; FGFR3:TACC3-Intron; FGFR3:TACC3V1; FGFR3:TACC3V3; or a combination thereof; and administering to the patient a pharmaceutically effective amount of an FGFR inhibitor if the one or more FGFR variants are present in the sample, wherein the FGFR inhibitor is the compound of formula (I):
- or a pharmaceutically acceptable salt thereof.
2. The method of claim 1, wherein the evaluating step further comprises measuring an expression level of PD-L1 in a biological sample and wherein the second administering step comprises administering the FGFR inhibitor if:
- the biological sample has a PD-L1 expression corresponding to an H-score of about 0 to about 99; or
- the biological sample has a PD-L1 expression level that is lower than a reference PD-L1 expression level.
3. The method of claim 1 or 2, wherein the biological sample is blood, lymph fluid, bone marrow, a solid tumor sample, or any combination thereof.
4. The method of claim 1, wherein the cancer is lung cancer, bladder cancer, gastric cancer, breast cancer, ovarian cancer, head and neck cancer, esophageal cancer, glioblastoma, or any combination thereof.
5. The method of claim 4, wherein the lung cancer is non-small cell lung cancer (NSCLC) adenocarcinoma, NSCLC squamous cell carcinoma, small cell lung cancer, or any combination thereof.
6. The method of claim 1, wherein the one or more FGFR variants further comprise an FGFR mutation, an FGFR amplification, or a combination thereof.
7. The method of claim 1, wherein the antibody that blocks an interaction between PD-1 and PD-L1 is an anti-PD-1 antibody, an anti-PD-L1 antibody, or a combination thereof.
8. The method of claim 2, wherein the cancer is lung cancer, bladder cancer, gastric cancer, breast cancer, ovarian cancer, head and neck cancer, esophageal cancer, glioblastoma, or any combination thereof.
9. The method of claim 8, wherein the lung cancer is non-small cell lung cancer (NSCLC) adenocarcinoma, NSCLC squamous cell carcinoma, small cell lung cancer, or any combination thereof.
10. The method of claim 2, wherein the one or more FGFR variants further comprise an FGFR mutation, an FGFR amplification, or a combination thereof.
11. The method of claim 2, wherein the antibody that blocks an interaction between PD-1 and PD-L1 is an anti-PD-1 antibody, an anti-PD-L1 antibody, or a combination thereof.
12. The method of claim 3, wherein the cancer is lung cancer, bladder cancer, gastric cancer, breast cancer, ovarian cancer, head and neck cancer, esophageal cancer, glioblastoma, or any combination thereof.
13. The method of claim 12, wherein the lung cancer is non-small cell lung cancer (NSCLC) adenocarcinoma, NSCLC squamous cell carcinoma, small cell lung cancer, or any combination thereof.
14. The method of claim 3, wherein the one or more FGFR variants further comprise an FGFR mutation, an FGFR amplification, or a combination thereof.
15. The method of claim 3, wherein the antibody that blocks an interaction between PD-1 and PD-L1 is an anti-PD-1 antibody, an anti-PD-L1 antibody, or a combination thereof.
16. The method of claim 1, wherein the biological sample:
- has a PD-L1 expression corresponding to an H-score of less than 20; or
- has a PD-L1 expression level that is lower than a reference PD-L1 expression level.
17. The method of claim 16, where the biological sample has a PD-L1 expression corresponding to an H-score of less than 20.
2940972 | June 1960 | Roch |
4666828 | May 19, 1987 | Gusella |
4683202 | July 28, 1987 | Mullis |
4801531 | January 31, 1989 | Frossard |
5192659 | March 9, 1993 | Simons |
5272057 | December 21, 1993 | Smulson et al. |
5700823 | December 23, 1997 | Hirth et al. |
5882864 | March 16, 1999 | An et al. |
6218529 | April 17, 2001 | An et al. |
6271231 | August 7, 2001 | Bergstrand et al. |
6331555 | December 18, 2001 | Hirth et al. |
7135311 | November 14, 2006 | David et al. |
7432279 | October 7, 2008 | Green et al. |
8409577 | April 2, 2013 | Thompson et al. |
8895601 | November 25, 2014 | Saxty et al. |
9067998 | June 30, 2015 | Clube |
9145367 | September 29, 2015 | Tazi et al. |
9221804 | December 29, 2015 | Leonard et al. |
9290478 | March 22, 2016 | Saxty et al. |
9303029 | April 5, 2016 | Woodhead et al. |
9303030 | April 5, 2016 | Angibaud |
9309241 | April 12, 2016 | Angibaud et al. |
9309242 | April 12, 2016 | Berdini et al. |
9399028 | July 26, 2016 | Tavazoie et al. |
9439896 | September 13, 2016 | Berdini et al. |
9447098 | September 20, 2016 | Saxty et al. |
9464071 | October 11, 2016 | Saxty |
9493426 | November 15, 2016 | Angibaud |
9527844 | December 27, 2016 | Angibaud et al. |
9737544 | August 22, 2017 | Angibaud |
9757364 | September 12, 2017 | Angibaud |
9850228 | December 26, 2017 | Saxty et al. |
9856236 | January 2, 2018 | Saxty et al. |
9902714 | February 27, 2018 | Vermeulen |
10039759 | August 7, 2018 | Berdini et al. |
10045982 | August 14, 2018 | Berdini et al. |
10052320 | August 21, 2018 | Woodhead et al. |
10085982 | October 2, 2018 | Jovcheva et al. |
20030207886 | November 6, 2003 | Plücker et al. |
20050261307 | November 24, 2005 | Cai et al. |
20050272728 | December 8, 2005 | Altenbach et al. |
20050272736 | December 8, 2005 | Altenbach et al. |
20070123494 | May 31, 2007 | Seipelt et al. |
20070149484 | June 28, 2007 | Claus et al. |
20080116789 | May 22, 2008 | Yamaguchi et al. |
20090054304 | February 26, 2009 | Herbert et al. |
20090118261 | May 7, 2009 | Aquila et al. |
20090221591 | September 3, 2009 | Hartmann et al. |
20090263397 | October 22, 2009 | Buck et al. |
20100228026 | September 9, 2010 | Yoshida et al. |
20100234347 | September 16, 2010 | Dollinger et al. |
20110123545 | May 26, 2011 | Marsh et al. |
20120302572 | November 29, 2012 | Kan et al. |
20130072457 | March 21, 2013 | Saxty |
20130096021 | April 18, 2013 | Chinnaiyan et al. |
20130267525 | October 10, 2013 | Saxty et al. |
20130296326 | November 7, 2013 | Pollock |
20140037642 | February 6, 2014 | McCaffery et al. |
20140288053 | September 25, 2014 | Berdini et al. |
20140296236 | October 2, 2014 | Berdini et al. |
20150017637 | January 15, 2015 | Chinnaiyan et al. |
20150031669 | January 29, 2015 | Woodhead et al. |
20150031703 | January 29, 2015 | Suzuki |
20150057293 | February 26, 2015 | Angibaud et al. |
20150086584 | March 26, 2015 | Gilboa et al. |
20150105368 | April 16, 2015 | Saxty et al. |
20150183875 | July 2, 2015 | Cobbold et al. |
20150191791 | July 9, 2015 | Shibata |
20150203589 | July 23, 2015 | Iavarone |
20150210769 | July 30, 2015 | Freeman |
20150239883 | August 27, 2015 | Angibaud et al. |
20150291589 | October 15, 2015 | Saxty et al. |
20150307945 | October 29, 2015 | Nakanishi et al. |
20160031856 | February 4, 2016 | Saxty |
20160031990 | February 4, 2016 | Steele et al. |
20160067336 | March 10, 2016 | Fandi et al. |
20160075666 | March 17, 2016 | Angibaud et al. |
20160090633 | March 31, 2016 | Platero |
20160108034 | April 21, 2016 | Angibaud et al. |
20160122410 | May 5, 2016 | Behrens et al. |
20160213677 | July 28, 2016 | Angibaud et al. |
20160220564 | August 4, 2016 | Woodhead et al. |
20160235744 | August 18, 2016 | Berdini et al. |
20160243228 | August 25, 2016 | Holash et al. |
20160287699 | October 6, 2016 | Karkera et al. |
20160311800 | October 27, 2016 | Saxty et al. |
20160347836 | December 1, 2016 | Grosso |
20170000781 | January 5, 2017 | Berdini et al. |
20170000796 | January 5, 2017 | Saxty et al. |
20170021019 | January 26, 2017 | Zibelman et al. |
20170100406 | April 13, 2017 | Jovcheva et al. |
20170101396 | April 13, 2017 | Vermeulen et al. |
20170105978 | April 20, 2017 | Angibaud et al. |
20170119763 | May 4, 2017 | Jovcheva et al. |
20170145102 | May 25, 2017 | Pierce et al. |
20170145103 | May 25, 2017 | Pierce et al. |
20180021332 | January 25, 2018 | Broggini |
20180127397 | May 10, 2018 | Saxty et al. |
20180186775 | July 5, 2018 | Vermeulen et al. |
20180296558 | October 18, 2018 | Jovcheva et al. |
2524525 | December 2004 | CA |
2524948 | December 2004 | CA |
1128496 | August 1996 | CN |
102036963 | April 2011 | CN |
0544445 | June 1993 | EP |
1001946 | May 2000 | EP |
1659175 | May 2006 | EP |
1208231 | January 2007 | EP |
1964837 | September 2008 | EP |
1990342 | November 2008 | EP |
2332939 | June 2011 | EP |
2650293 | October 2013 | EP |
3027210 | June 2016 | EP |
3177321 | June 2017 | EP |
3179992 | June 2017 | EP |
2003213463 | July 2003 | JP |
2006516561 | July 2006 | JP |
2008530030 | August 2008 | JP |
2008540535 | November 2008 | JP |
2010514693 | May 2010 | JP |
2377241 | December 2009 | RU |
94/26723 | November 1994 | WO |
95/19169 | July 1995 | WO |
98/54156 | December 1998 | WO |
99/17759 | April 1999 | WO |
00/42026 | July 2000 | WO |
01/19825 | February 2001 | WO |
01/68047 | September 2001 | WO |
02/076985 | October 2002 | WO |
03/051833 | June 2003 | WO |
03/055491 | July 2003 | WO |
03/086394 | October 2003 | WO |
2004/006355 | January 2004 | WO |
2004/030635 | April 2004 | WO |
2004/043950 | May 2004 | WO |
2004/056822 | July 2004 | WO |
2004065378 | August 2004 | WO |
2004/098494 | November 2004 | WO |
2004/110350 | December 2004 | WO |
2005/007099 | January 2005 | WO |
2005/009437 | February 2005 | WO |
2005/012288 | February 2005 | WO |
2005/039587 | May 2005 | WO |
2005/047244 | May 2005 | WO |
2005/054201 | June 2005 | WO |
2005054231 | June 2005 | WO |
2005/061463 | July 2005 | WO |
2006/040052 | April 2006 | WO |
2006/066361 | June 2006 | WO |
2006084338 | August 2006 | WO |
2006/092430 | September 2006 | WO |
2006121168 | November 2006 | WO |
2006124354 | November 2006 | WO |
2007/003419 | January 2007 | WO |
2007005874 | January 2007 | WO |
2007/023186 | March 2007 | WO |
2007054556 | May 2007 | WO |
2007/075567 | July 2007 | WO |
2007/125405 | November 2007 | WO |
2007/132227 | November 2007 | WO |
2008/003702 | January 2008 | WO |
2008060907 | May 2008 | WO |
2008/076278 | June 2008 | WO |
2008/078091 | July 2008 | WO |
2008/082198 | July 2008 | WO |
2008079988 | July 2008 | WO |
2008080015 | July 2008 | WO |
2008109465 | September 2008 | WO |
2008112408 | September 2008 | WO |
2008/138878 | November 2008 | WO |
2008/141065 | November 2008 | WO |
2008/148867 | December 2008 | WO |
2008/150827 | December 2008 | WO |
2008/155378 | December 2008 | WO |
2009/019518 | February 2009 | WO |
2009/021083 | February 2009 | WO |
2009020990 | February 2009 | WO |
2009/064835 | May 2009 | WO |
2009/137378 | November 2009 | WO |
2009/141386 | November 2009 | WO |
2010059771 | May 2010 | WO |
2010/084152 | July 2010 | WO |
2010088177 | August 2010 | WO |
2010129570 | November 2010 | WO |
2011/026579 | March 2011 | WO |
2011/028947 | March 2011 | WO |
2011/064250 | June 2011 | WO |
2011/126903 | October 2011 | WO |
2011/135376 | November 2011 | WO |
2011/146591 | November 2011 | WO |
2011/149937 | December 2011 | WO |
2012/073017 | June 2012 | WO |
2012/104776 | August 2012 | WO |
2012106556 | August 2012 | WO |
2012/118492 | September 2012 | WO |
2012/148540 | November 2012 | WO |
2013019906 | February 2013 | WO |
2013/032951 | March 2013 | WO |
2013/040515 | March 2013 | WO |
2013/043935 | March 2013 | WO |
2013/052699 | April 2013 | WO |
2013/061074 | May 2013 | WO |
2013/061077 | May 2013 | WO |
2013/061080 | May 2013 | WO |
2013/061081 | May 2013 | WO |
2013/061305 | May 2013 | WO |
2013/063217 | May 2013 | WO |
2013076186 | May 2013 | WO |
2013087725 | June 2013 | WO |
2013089882 | June 2013 | WO |
2013133351 | September 2013 | WO |
2013173485 | November 2013 | WO |
2013/179033 | December 2013 | WO |
2013/179034 | December 2013 | WO |
2014007369 | January 2014 | WO |
2014018673 | January 2014 | WO |
2014018841 | January 2014 | WO |
2014051022 | April 2014 | WO |
2014071419 | May 2014 | WO |
2014113729 | July 2014 | WO |
2014/165422 | October 2014 | WO |
2014/174307 | October 2014 | WO |
2014165710 | October 2014 | WO |
2014193229 | December 2014 | WO |
2015/016718 | February 2015 | WO |
2015017607 | February 2015 | WO |
2015/077717 | May 2015 | WO |
2015/100257 | July 2015 | WO |
2015/112900 | July 2015 | WO |
2015144803 | October 2015 | WO |
2015144804 | October 2015 | WO |
2015144808 | October 2015 | WO |
2016/004218 | January 2016 | WO |
2016/004875 | January 2016 | WO |
2016/019472 | February 2016 | WO |
2016/024228 | February 2016 | WO |
2016/024231 | February 2016 | WO |
2016/040880 | March 2016 | WO |
2016/040882 | March 2016 | WO |
2016/044207 | March 2016 | WO |
2016048833 | March 2016 | WO |
2016/054555 | April 2016 | WO |
2016/061142 | April 2016 | WO |
2016/065409 | May 2016 | WO |
2016/094309 | June 2016 | WO |
2016/100882 | June 2016 | WO |
2016/118654 | July 2016 | WO |
2016/128912 | August 2016 | WO |
2016128411 | August 2016 | WO |
2016134234 | August 2016 | WO |
2016/137850 | September 2016 | WO |
2016/140717 | September 2016 | WO |
2016/141209 | September 2016 | WO |
2016/141218 | September 2016 | WO |
2016/153839 | September 2016 | WO |
2016/154068 | September 2016 | WO |
2016/154473 | September 2016 | WO |
2016/161239 | October 2016 | WO |
2016/168716 | October 2016 | WO |
2016/191751 | December 2016 | WO |
2016/196389 | December 2016 | WO |
2016/201425 | December 2016 | WO |
2016/210108 | December 2016 | WO |
2017/004192 | January 2017 | WO |
2017/013436 | January 2017 | WO |
2017/046746 | March 2017 | WO |
2017/091577 | June 2017 | WO |
2017/091580 | June 2017 | WO |
2017/093942 | June 2017 | WO |
- International Search Report for PCT/US2016/025482 dated Aug. 5, 2016.
- Moreira Da Silva, R., “Novolumab: Anti-PD-1 monoclonal antibody cancer immunotherapy”, Drugs of the Future, vol. 39, No. 1, pp. 15-24 (2014).
- Ho, H.K., et al., “Current strategies for inhibiting FGFR activities in clinical applications: opportunities, challenges and toxicological considerations”, Drug Discovery Today, vol. 19, Issue 1, Abstract only (2014).
- Yan, L., et al., “An efficient synthesis of quinoxaline derivatives from 4-chloro-4-deoxy-α-D-galactose and their cytotoxic activities”, Bioorganic & Medicinal Chemistry Letters, vol. 17, No. 3, 2006, pp. 609-612.
- Thompson, A.M., et al. “Synthesis and Structure—Activity Relationships of 7-Substituted 3-(2,6-Dichlorophenyl)-1,6-napthyridin-2(1H)-ones as Selective Inhibitors of pp60c-src”, Journal of Medicinal Chemistry, vol. 43, No. 16, 2000, pp. 3134-3147.
- Berge, S.M., et al. “Pharmaceutical Salts”, Journal of Pharmaceutical Sciences, vol. 66, No. 1, 1977, pp. 1-19.
- Deady, L.W. “Ring Nitrogen Oxidation of Amino Substituted Nitrogen Heterocycles with m-Chloroperbenzoic Acid”, Synthetic Communications, vol. 7(8), 1977, pp. 509-514.
- Knights, V., et al. “De-regulated FGF receptors as therapeutic targets in cancer”, Pharmacology & Therapeutics, 2010; vol. 125(1), pp. 105-117.
- Korc, M., et al. “The Role of Fibroblast Growth Factors in Tumor Growth”, Current Cancer Drug Targets, vol. 9(5), 2009, pp. 639-651.
- Angerer, L.M., et al. “Demonstration of Tissue-Specific Gene Expression by in Situ Hybridization”, Methods in Enzymology, vol. 152, 1987, pp. 649-661.
- Deprimo, S.E., et al. “Expression profiling of blood samples from an SU5416 Phase III metastatic colorectal cancer clinical trial: a novel strategy for biomarker identification”, BMC Cancer, vol. 3, 2003; pp. 1-12.
- Orre, M., et al., “VEGF, VEGFR-1, VEGFR-2, Microvessel Density and Endothelial Cell Proliferation in Tumours of the Ovary”, Int. J. Cancer (Pred. Oncol.), vol. 84(2), 1999, pp. 101-108.
- Zhou, W., et al., “A Structure-Guided Approach to Creating Covalent FGFR Inhibitors”, Chemistry & Biology, vol. 17, pp. 285-295 (2010).
- Avendaño, C., et al., “Drugs That Inhibit Signalling Pathways for Tumor Cell Growth and Proliferation”, Medicinal Chemistry of Anticancer Drugs, pp. 251-305 (2008).
- Garuti, L., et al., Irreversible Protein Kinase Inhibitors, Current Medicinal Chemistry, vol. 18, No. 20, Jul. 1, 2011, pp. 2981-2994.
- Vippagunta, S.R. et al., “Crystalline Solids”, Advanced Drug Delivery Reviews, vol. 48, pp. 3-26 (2001).
- Jordan, V.C., “Tamoxifen: A Most Unlikely Pioneering Medicine”, Nature Reviews: Drug Discovery, vol. 2, pp. 205-213 (2003).
- Hackam, D.G., et al., “Translation of Research Evidence From Animals to Humans”, JAMA, vol. 14, pp. 1731-1732 (2006).
- “Himicheskaja jenciklopedija” tom 4, str. 990-993, izdatel'stvo “Sovetskaja jencklopedija”, Moskva, 1988 (In English: Chemical Encyclopedia, vol. 4, pp. 990-993, Publishing House “Soviet encyclopedia”, Moscow, 1988).
- V. Hikkinvottom, “Reakcii Organicheskih Soedinenij” Gosudarstvennoe ob#eninennoe nauchno-technicheskoe izdatel'stvo, Redakcija himicheskoj literatury, Moskva, stranicy 360-362, 1939 (In English: V. Hikkinbottom, “Reactions of Organic Compounds”, State Associated Scientific-Technical Publishing House, Editor Office of Chemical Literature, pp. 360-362, Moscow, 1939).
- “Himicheskaja jenciklopedija” tom. 1, stranicy 242-243, izdatel'stvo “Sovetskaja jencklopedija”, Moskva, 1988 (In English: Chemical Encyclopedia (thesaurus), vol. 1, pp. 242-243, publishing house “Soviet encyclopedia”, Moscow, 1988).
- Dorwald, F.Z., “Side Reactions in Organic Synthesis: A Guide to Successful Synthesis Design”, Weinheim:WILEY-VCH Verlag GmbH & Co. KGaA, 2005, ISBN: 3-527-31021.5.
- Lima, L.M., et al., “Bioisosterism: A Useful Strategy for Molecular Modification and Drug Design”, Current Medical Chemistry, vol. 12(1), pp. 23-49 (2005).
- Patani, G.A., et al., “Bioisosterism: A Rational Approach in Drug Design”, Chem. Rev. vol. 96, pp. 3147-3176 (1996).
- Dieci, M.V., et al., “Fibroblast Growth Factor Receptor Inhibitors as a Cancer Treatment: From a Biologic Rationale to Medical Perspectives”, Cancer Discovery, vol. 3, No. 3, pp. 264-279 (Feb. 2013).
- Gallick, G.E., et al., “Small-molecule protein tyrosine kinase inhibitors for the treatment of metastatic prostate cancer”, Future Medicinal Chemistry, vol. 4, No. 1, pp. 107-119 (Jan. 2012).
- Study to Assess the Relative Bioavailability of Orally Administered JNJ-42756493 Tablet Versus JNJ-42756493 Capsule in Healthy Participants, ClinicalTrials.gov, pp. 1-4 (2014).
- Matsuda, Y., et al., “Fibroblast Growth Factor Receptor-2 IIIc as a Novel Molecular Target in Colorectal Cancer”, Current Colorectal Cancer Reports, vol. 10, No. 1, pp. 20-26 (2014).
- Carneiro, B.A., et al., “Emerging therapeutic targets in bladder cancer”, Cancer Treatment Reviews, vol. 41, No. 2, pp. 170-178 (2015).
- Fujita, M., et al., “Generation of Formaldehyde by Pharmaceutical Excipients and Its Absorption by Meglumine”, Chem. Pharm. Bull, vol. 57, No. 10, pp. 1096-1099 (2009).
- Adcock, J., et al., “Diversity oriented synthesis: substitution at C5 in unreactive pyrimidines by Claisen rearrangement and reactivity in nucleophilic substitution at C2 and C4 in pteridines and pyrido[2,3-d]pyrimidines”, Tetrahedron, vol. 67, pp. 3226-3237 (2011).
- Database Caplus, Grina, et al., “Preparation of oxohydroquinazolinylaminophenylpropanesulfonamide derivatives and analogs for use as Raf inhibitors”, Document No. 157:465574, Accession No. 2012:1301209 (2012).
- Liang, G., et al., “Small molecule inhibition of fibroblast growth factor receptors in cancer”, Cytokine & Growth Factor Reviews, vol. 24, pp. 467-475 (2013).
- Golub, T.R., et al., “Molecular Classification of Cancer: Class Discovery and Class Prediction by Gene Expression Monitoring”, Science, vol. 286, pp. 531-537 (1999).
- Greulich, H., et al., “Targeting mutant fibroblast growth factor receptors in cancer”, Trends in Molecular Medicine, vol. 17, No. 5, pp. 283-292 (2011).
- Freshney, R.I., “Culture of Animal Cells, A Manual of Basic Technique”, Published by Alan R. Liss, Inc, New York, pp. 1-6 (1983).
- Cohen, P., “The development and therapeutic potential of protein kinase inhibitors”, Current Opinion in Chemical Biology, vol. 3, pp. 459-465 (1999).
- Cecil Textbook of Medicine, edited by Bennet, J.C., and Plum, F., 20th edition, vol. 1, pp. 1004-1010 (1996).
- Hynes, N.E., et al., “Potential for Targeting the Fibroblast Growth Factor Receptors in Breast Cancer”, Cancer Research, vol. 70, pp. 5199-5202 (2010).
- Neidle, S., “Cancer Drug Design and Discovery”, Elsevier/Academic Press, pp. 427-431 (2008).
- Dermer, G.B., “Another Anniversary for the War on Cancer”, Biotechnology, vol. 12, p. 320 (1994).
- Katoh, Y., et al., “FGFR2-related pathogenesis and FGFR2-targeted therapeutics (Review)”, International Journal of Molecular Medicine, vol. 23, pp. 307-311 (2009).
- Jain, V.K., et al., “Challenges and opportunities in the targeting of fibroblast growth factor receptors in breast cancer”, Breast Cancer Research, vol. 14, No. 208, pp. 1-9 (2012).
- Sonpavde, G., et al., “Fibroblast growth factor receptors as therapeutic targets in clear-cell renal cell carcinoma”, Expert Opinion on Investigational Drugs, vol. 23, Issue 3, pp. 305-315 (2014).
- Rodriguez-Vida, A., et al., “Complexity of FGFR signaling in metastatic urothelial cancer,” Journal of Hematology & Oncology, vol. 8, No., pp. 119 et seq. (2015).
- Angibaud et al., “Discovery of JNJ-42756493, A Potent Fibroblast Growth Factor Receptor (FGFR) Inhibitor Using a Fragment Based Approach,” AACR Minisymposium, Small Molecule Design and Optimization San Diego, CA, Apr. 8, 2014, 16 pp.
- Bronte et al., “Nintedanib in NSCLC: Evidence to Date and Place in Therapy,” Therapeutic Advances in Medical Oncology, 2016, vol. 8[3], pp. 188-197.
- Kathoh et al., “FGFR inhibitors: Effects on Cancer Cells, Tumor Microenvironment and Whole-Body Homeostasis (Review),” International Journal of Molecular Medicine 2016, 38(1), pp. 3-15.
- D.A.Kharkevich, Farmakologiya (Pharmacology), 1996, M., Meditsina, p. 41, chapter 6.A (in Russian Only).
- V.G.Belikov, Farmatsevticheskaya khimiya (Pharmaceutical Chemistry), M., Vysshaya shkola, 1993, p. 1, chapter 2.2, pp. 43-47) (in Russian only).
- Amin et al., “Nivolumab (anti-PD-1; BMS-936558, ONO-4538) in combination with sunitinib or pazopanib in patients (pts) with metastic renal cell carcinoma (mRCC),” Journal of Clinical Oncology 2014, vol. 32:15.
- Phillips et al., “Therapeutic Uses of Anti-PD-1 and anti-PD-L1 Antibodies,” International Immunology, Oct. 2014, vol. 27, No. 1, pp. 39-46.
- Dienstmann et al., “Genomic Aberrations in the FGFR Pathway: Opportunities for Targeted Therapies in Solid Tumors,” Annals of Oncology, vol. 25, Nov. 20, 2013, No. 3, pp. 552-563.
- Parker et al., “Emergence of FGFR Family Gene Fusions as Therapeutic Targets in a Wide Spectrum of Solid Tumours,” Journal of Pathology, Oct. 29, 2013, vol. 232, No. 1, pp. 4-15.
- Arai et al., “Fibroblast Growth Factor Receptor 2 Tyrosine Kinase Fusions Define a Unique Molecular Subtype of Cholangiocarcinoma,” Hepatology, Apr. 2014, vol. 59, No. 4, pp. 1427-1434.
- Bahleda et al., “Phase 1 Study of JNJ-42756493, a Pan-Fibroblast Growth Factor Receptor (FGFR) Inhibitor, in patients with advanced Solid Tumors,” Journal of Clinical Oncology, May 2014, vol. 32, No. 15, pp. 2501-2501.
- Di Siefano et al., “Detection, Characterization, and Inhibition of FGFR-TACC Fusions in IDH Wild-Type Glioma,” Clinical Cancer Research, Jan. 21, 2015, vol. 21, No. 14, pp. 3307-3317.
- Parker, B.C., et al., “The tumorigenic FGFR3-TACC3 gene fusion escapes miR-99a regulation in glioblastoma,” The Journal of Clinical Investigation, 123 (2), pp. 855-865, Feb. 1, 2013.
- Bello, et al., “E=3810 is a Potent Dual Inhibitor of VEGFR and FGFR that Exerts Antitumor Activity in Multiple Preclinical Models,” vol. 71(4), pp. 1396-1405 (2011).
- Database, Geneseq [Online], “FGFR3-TACC3 gene fusion PCR primer, FGFR3-TACC3(F18T11)_qPCR_F SEQ: 15,” XP002753027, Database accession No. BAT14432 (2013).
- Database, Geneseq [Online], “Human FGFR 2 mRNA target sequence for mdRNA, SEQ ID:3954,” XP055257043, Database accession no. ATM46802 (2008).
- Gavine, et al., “AZD4547: An Orally Bioavailable, Potent, and Selective Inhibitor of the Fibroblast Growth Factor Receptor Tyrosine Kinase Family,” Cancer Research, vol. 72(8), pp. 2045-2056 (2012).
- International Search Report from PCT/US2015/050996 dated Mar. 23, 2016.
- Mengual, et al., BMC Research Notes 1:21, pp. 1-8 (Jun. 2008).
- Millholland, et al., Research and Reports in Urology, 4: 33-40 (2012).
- Sabnis, et al., “FGFR Fusions in the Driver's Seat,” Cancer Discovery, vol. 3 (6), pp. 607-609 (2013).
- Shinmura, et al., “A novel somatic FGFR3 mutation in primary lung cancer,” Oncology Reports, vol. 31 (3), pp. (2014).
- Singh, et al., “Transforming Fusions of FGFR and TACC Genes in Human Glioblastoma, ” Science, vol. 337 (6099), pp. 1231-1235 (2012).
- Trudel, et al., “Evaluation of XL999, a Potent Inhibitor of FGFR3, for the Potential Treatment of t(4;14) Positive Multiple Myeloma, ” Blood, vol. 110 (11), pp. 741A-742A (2007).
- Williams, et al., “Oncogenic FGFR3 gene fusions in bladder cancer,” Human Molecular Genetics, vol. 22 (4), (2013). pp. 795-803.
- Wu, et al., “Identification of Targetable FGFR Gene Fusions in Diverse Cancers,” Cancer Discovery, vol. 3 (6), pp. 636-647 (2013).
Type: Grant
Filed: Mar 24, 2016
Date of Patent: Nov 19, 2019
Patent Publication Number: 20160287699
Assignee: ASTEX THERAPEUTICS LTD (Cambridge)
Inventors: Jayaprakash Karkera (Germantown, MD), Suso Jesus Platero (Washington Crossing, PA), Raluca Verona (Swarthmore, PA), Matthew V. Lorenzi (Philadelphia, PA)
Primary Examiner: Peter J Reddig
Application Number: 15/079,136
International Classification: A61K 45/06 (20060101); C07K 16/28 (20060101); A61K 31/498 (20060101); A61K 39/395 (20060101); C12Q 1/6886 (20180101); G01N 33/574 (20060101);